Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(30): e2121267119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35867829

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) affects more than 500,000 individuals in the United States alone. In most cases, ADPKD is caused by a loss-of-function mutation in the PKD1 gene, which encodes polycystin-1 (PC1). Previous studies reported that PC1 interacts with atypical protein kinase C (aPKC). Here we show that PC1 binds to the ζ isoform of aPKC (PKCζ) and identify two PKCζ phosphorylation sites on PC1's C-terminal tail. PKCζ expression is down-regulated in patients with ADPKD and orthologous and nonorthologous PKD mouse models. We find that the US Food and Drug Administration-approved drug FTY720 restores PKCζ expression in in vitro and in vivo models of polycystic kidney disease (PKD) and this correlates with ameliorated disease progression in multiple PKD mouse models. Importantly, we show that FTY720 treatment is less effective in PKCζ null versions of these PKD mouse models, elucidating a PKCζ-specific mechanism of action that includes inhibiting STAT3 activity and cyst-lining cell proliferation. Taken together, our results reveal that PKCζ down-regulation is a hallmark of PKD and that its stabilization by FTY720 may represent a therapeutic approach to the treat the disease.


Asunto(s)
Clorhidrato de Fingolimod , Riñón Poliquístico Autosómico Dominante , Proteína Quinasa C , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Activación Enzimática , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Ratones , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/enzimología , Proteína Quinasa C/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
2.
Am J Physiol Renal Physiol ; 326(4): F644-F660, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38420674

RESUMEN

Patients with hypertension or obesity can develop glomerular dysfunction characterized by injury and depletion of podocytes. To better understand the molecular processes involved, young mice were treated with either deoxycorticosterone acetate (DOCA) or fed a high-fat diet (HFD) to induce hypertension or obesity, respectively. The transcriptional changes associated with these phenotypes were measured by unbiased bulk mRNA sequencing of isolated podocytes from experimental models and their respective controls. Key findings were validated by immunostaining. In addition to a decrease in canonical proteins and reduced podocyte number, podocytes from both hypertensive and obese mice exhibited a sterile inflammatory phenotype characterized by increases in NLR family pyrin domain containing 3 (NLRP3) inflammasome, protein cell death-1, and Toll-like receptor pathways. Finally, although the mice were young, podocytes in both models exhibited increased expression of senescence and aging genes, including genes consistent with a senescence-associated secretory phenotype. However, there were differences between the hypertension- and obesity-associated senescence phenotypes. Both show stress-induced podocyte senescence characterized by increased p21 and p53. Moreover, in hypertensive mice, this is superimposed upon age-associated podocyte senescence characterized by increased p16 and p19. These results suggest that senescence, aging, and inflammation are critical aspects of the podocyte phenotype in experimental hypertension and obesity in mice.NEW & NOTEWORTHY Hypertension and obesity can lead to glomerular dysfunction in patients, causing podocyte injury and depletion. Here, young mice given deoxycorticosterone acetate or a high-fat diet to induce hypertension or obesity, respectively. mRNA sequencing of isolated podocytes showed transcriptional changes consistent with senescence, a senescent-associated secretory phenotype, and aging, which was confirmed by immunostaining. Ongoing studies are determining the mechanistic roles of the accelerated aging podocyte phenotype in experimental hypertension and obesity.


Asunto(s)
Hipertensión , Enfermedades Renales , Podocitos , Humanos , Ratones , Animales , Anciano , Podocitos/metabolismo , Ratones Obesos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Fenotipo , Enfermedades Renales/metabolismo , Obesidad/metabolismo , Hipertensión/genética , Hipertensión/metabolismo , Desoxicorticosterona , Acetatos/metabolismo , ARN Mensajero/metabolismo
3.
Am J Physiol Renal Physiol ; 326(1): F120-F134, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37855038

RESUMEN

As life expectancy continues to rise, age-related diseases are becoming more prevalent. For example, proteinuric glomerular diseases typified by podocyte injury have worse outcomes in the elderly compared with young patients. However, the reasons are not well understood. We hypothesized that injury to nonaged podocytes induces senescence, which in turn augments their aging processes. In primary cultured human podocytes, injury induced by a cytopathic antipodocyte antibody, adriamycin, or puromycin aminonucleoside increased the senescence-related genes CDKN2A (p16INK4a/p14ARF), CDKN2D (p19INK4d), and CDKN1A (p21). Podocyte injury in human kidney organoids was accompanied by increased expression of CDKN2A, CDKN2D, and CDKN1A. In young mice, experimental focal segmental glomerulosclerosis (FSGS) induced by adriamycin and antipodocyte antibody increased the glomerular expression of p16, p21, and senescence-associated ß-galactosidase (SA-ß-gal). To assess the long-term effects of early podocyte injury-induced senescence, we temporally followed young mice with experimental FSGS through adulthood (12 m of age) and middle age (18 m of age). p16 and Sudan black staining were higher at middle age in mice with earlier FSGS compared with age-matched mice that did not get FSGS when young. This was accompanied by lower podocyte density, reduced canonical podocyte protein expression, and increased glomerular scarring. These results are consistent with injury-induced senescence in young podocytes, leading to increased senescence of podocytes by middle age accompanied by lower podocyte lifespan and health span.NEW & NOTEWORTHY Glomerular function is decreased by aging. However, little is known about the molecular mechanisms involved in age-related glomerular changes and which factors could contribute to a worse glomerular aging process. Here, we reported that podocyte injury in young mice and culture podocytes induced senescence, a marker of aging, and accelerates glomerular aging when compared with healthy aging mice.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Enfermedades Renales , Podocitos , Persona de Mediana Edad , Humanos , Ratones , Animales , Anciano , Podocitos/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomérulos Renales/metabolismo , Enfermedades Renales/metabolismo , Envejecimiento , Doxorrubicina/toxicidad , Doxorrubicina/metabolismo
4.
Kidney Int ; 104(3): 455-462, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37290603

RESUMEN

The majority of podocyte disorders are progressive in nature leading to chronic kidney disease and often kidney failure. The scope of current therapies is typically nonspecific immunosuppressant medications, which are accompanied by unwanted and serious side effects. However, many exciting clinical trials are underway to reduce the burden of podocyte diseases in our patients. Major advances and discoveries have recently been made experimentally in our understanding of the molecular and cellular mechanisms underlying podocyte injury in disease. This begs the question of how best to take advantage of these impressive strides. One approach to consider is the repurposing of therapeutics that have already been approved by the Food and Drug Administration, European Medicines Agency, and other regulatory agencies for indications beyond the kidney. The advantages of therapy repurposing include known safety profiles, drug development that has already been completed, and overall reduced costs for studying alternative indications for selected therapies. The purpose of this mini review is to examine the experimental literature of podocyte damage and determine if there are mechanistic targets in which prior approved therapies can be considered for repurposing to podocyte disorders.


Asunto(s)
Podocitos , Insuficiencia Renal Crónica , Humanos , Preparaciones Farmacéuticas , Reposicionamiento de Medicamentos , Riñón , Insuficiencia Renal Crónica/tratamiento farmacológico
5.
Kidney Int ; 102(1): 12-13, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35738826

RESUMEN

Regenerative repair following injury to proximal tubular epithelial cells (PTECs) is essential to restore the kidney to normal function in acute kidney injury. Failure to accomplish this leads to chronic kidney disease. Expression of the paired-box transcription factor Pax2 in PTECs is required for their regenerative proliferation and repair. However, a loss-of-function study now shows that the absence of Pax2 not only impacts PTEC proliferation but also causes myofibroblast recruitment leading to excessive tubulointerstitial fibrosis.


Asunto(s)
Lesión Renal Aguda , Factor de Transcripción PAX2 , Lesión Renal Aguda/patología , Animales , Células Epiteliales/metabolismo , Fibrosis , Riñón/metabolismo , Túbulos Renales Proximales/patología , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo
6.
Kidney Int ; 102(5): 966-968, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36272754

RESUMEN

Podocytes undergo defined morphologic changes during development, homeostasis, and aging, and on injury. Quantitative podometric assessments of podocyte endowment provide a powerful tool to interrogate glomerular health. Expanding this approach to a regional assessment demonstrates that the podocytes from cortical, subcortical, and juxtamedullary glomeruli are not only morphologically heterogeneous per se, but respond differently to stressors, such as age and hypertension. This suggests that zonal glomerular changes harbor critical information to understand glomerulopathies.


Asunto(s)
Hipertensión , Enfermedades Renales , Podocitos , Humanos , Glomérulos Renales
7.
J Am Soc Nephrol ; 32(11): 2697-2713, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34716239

RESUMEN

The effects of healthy aging on the kidney, and how these effects intersect with superimposed diseases, are highly relevant in the context of the population's increasing longevity. Age-associated changes to podocytes, which are terminally differentiated glomerular epithelial cells, adversely affect kidney health. This review discusses the molecular and cellular mechanisms underlying podocyte aging, how these mechanisms might be augmented by disease in the aged kidney, and approaches to mitigate progressive damage to podocytes. Furthermore, we address how biologic pathways such as those associated with cellular growth confound aging in humans and rodents.


Asunto(s)
Envejecimiento/fisiología , Podocitos/citología , Adulto , Anciano , Animales , Autofagia , Restricción Calórica , Ciclo Celular , Forma de la Célula , Células Cultivadas , Senescencia Celular , Daño del ADN , Femenino , Expresión Génica , Humanos , Inflamasomas , Glomérulos Renales/citología , Glomérulos Renales/crecimiento & desarrollo , Masculino , Ratones , Persona de Mediana Edad , Mitocondrias/metabolismo , Modelos Animales , Oligopéptidos/farmacología , Estrés Oxidativo , Podocitos/metabolismo , Ratas , Muerte Celular Regulada , Sirtuinas/metabolismo , Especificidad de la Especie , Adulto Joven
8.
Development ; 145(6)2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29530879

RESUMEN

The development of the kidney relies on the establishment and maintenance of a precise tubular diameter of its functional units, the nephrons. This process is disrupted in polycystic kidney disease (PKD), resulting in dilations of the nephron and renal cyst formation. In the course of exploring G-protein-coupled signaling in the Xenopus pronephric kidney, we discovered that loss of the G-protein α subunit, Gnas, results in a PKD phenotype. Polycystin 1, one of the genes mutated in human PKD, encodes a protein resembling a G-protein-coupled receptor. Furthermore, deletion of the G-protein-binding domain present in the intracellular C terminus of polycystin 1 impacts functionality. A comprehensive analysis of all the G-protein α subunits expressed in the Xenopus pronephric kidney demonstrates that polycystin 1 recruits a select subset of G-protein α subunits and that their knockdown - as in the case of Gnas - results in a PKD phenotype. Mechanistically, the phenotype is caused by increased endogenous G-protein ß/γ signaling and can be reversed by pharmacological inhibitors as well as knocking down Gnb1. Together, our data support the hypothesis that G proteins are recruited to the intracellular domain of PKD1 and that this interaction is crucial for its function in the kidney.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Riñón/metabolismo , Enfermedades Renales Poliquísticas/genética , Canales Catiónicos TRPP/metabolismo , Animales , Técnicas de Cultivo de Célula , Humanos , Hibridación in Situ , Enfermedades Renales Poliquísticas/metabolismo , Transducción de Señal , Resonancia por Plasmón de Superficie , Xenopus laevis/metabolismo
9.
Pediatr Res ; 89(1): 157-162, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32283547

RESUMEN

BACKGROUND: Autosomal recessive polycystic kidney disease (ARPKD) is a rare but potentially lethal genetic disorder typically characterized by diffuse renal microcysts. Clinical trials for patients with ARPKD are not currently possible due to the absence of sensitive measures of ARPKD kidney disease progression and/or therapeutic efficacy. METHODS: In this study, animal and human magnetic resonance imaging (MRI) scanners were used to obtain quantitative kidney T1 and T2 relaxation time maps for both excised kidneys from bpk and wild-type (WT) mice as well as for a pediatric patient with ARPKD and a healthy adult volunteer. RESULTS: Mean kidney T1 and T2 relaxation times showed significant increases with age (p < 0.05) as well as significant increases in comparison to WT mice (p < 2 × 10-10). Significant or nearly significant linear correlations were observed for mean kidney T1 (p = 0.030) and T2 (p = 0.054) as a function of total kidney volume, respectively. Initial magnetic resonance fingerprinting assessments in a patient with ARPKD showed visible increases in both kidney T1 and T2 in comparison to the healthy volunteer. CONCLUSIONS: These preclinical and initial clinical MRI studies suggest that renal T1 and T2 relaxometry may provide an additional outcome measure to assess cystic kidney disease progression in patients with ARPKD. IMPACT: A major roadblock for implementing clinical trials in patients with ARPKD is the absence of sensitive measures of ARPKD kidney disease progression and/or therapeutic efficacy. A clinical need exists to develop a safe and sensitive measure for kidney disease progression, and eventually therapeutic efficacy, for patients with ARPKD. Mean kidney T1 and T2 MRI relaxation times showed significant increases with age (p < 0.05) as well as significant increases in comparison to WT mice (p < 2 ×10-10), indicating that T1 and T2 may provide sensitive assessments of cystic changes associated with progressive ARPKD kidney disease. This preclinical and initial clinical study suggests that MRI-based kidney T1 and T2 mapping could be used as a non-invasive assessment of ARPKD kidney disease progression. These non-invasive, quantitative MRI techniques could eventually be used as an outcome measure for clinical trials evaluating novel therapeutics aimed at limiting or preventing ARPKD kidney disease progression.


Asunto(s)
Riñón/diagnóstico por imagen , Imagen por Resonancia Magnética , Riñón Poliquístico Autosómico Recesivo/diagnóstico por imagen , Adolescente , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Riñón Poliquístico Autosómico Recesivo/genética , Valor Predictivo de las Pruebas
11.
J Am Soc Nephrol ; 28(10): 2973-2984, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28620080

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is caused by inactivating mutations in PKD1 (85%) or PKD2 (15%). The ADPKD proteins encoded by these genes, polycystin-1 (PC1) and polycystin-2 (PC2), form a plasma membrane receptor-ion channel complex. However, the mechanisms controlling the subcellular localization of PC1 and PC2 are poorly understood. Here, we investigated the involvement of the retromer complex, an ancient protein module initially discovered in yeast that regulates the retrieval, sorting, and retrograde transport of membrane receptors. Using yeast two-hybrid, biochemical, and cellular assays, we determined that PC2 binds two isoforms of the retromer-associated protein sorting nexin 3 (SNX3), including a novel isoform that binds PC2 in a direct manner. Knockdown of SNX3 or the core retromer protein VPS35 increased the surface expression of endogenous PC1 and PC2 in vitro and in vivo and increased Wnt-activated PC2-dependent whole-cell currents. These findings indicate that an SNX3-retromer complex regulates the surface expression and function of PC1 and PC2. Molecular targeting of proteins involved in the endosomal sorting of PC1 and PC2 could lead to new therapeutic approaches in ADPKD.


Asunto(s)
Endocitosis , Nexinas de Clasificación/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Células HEK293 , Células HeLa , Humanos , Túbulos Renales/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Xenopus
12.
J Am Soc Nephrol ; 28(5): 1370-1378, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28096308

RESUMEN

(Re)Building a Kidney is a National Institute of Diabetes and Digestive and Kidney Diseases-led consortium to optimize approaches for the isolation, expansion, and differentiation of appropriate kidney cell types and the integration of these cells into complex structures that replicate human kidney function. The ultimate goals of the consortium are two-fold: to develop and implement strategies for in vitro engineering of replacement kidney tissue, and to devise strategies to stimulate regeneration of nephrons in situ to restore failing kidney function. Projects within the consortium will answer fundamental questions regarding human gene expression in the developing kidney, essential signaling crosstalk between distinct cell types of the developing kidney, how to derive the many cell types of the kidney through directed differentiation of human pluripotent stem cells, which bioengineering or scaffolding strategies have the most potential for kidney tissue formation, and basic parameters of the regenerative response to injury. As these projects progress, the consortium will incorporate systematic investigations in physiologic function of in vitro and in vivo differentiated kidney tissue, strategies for engraftment in experimental animals, and development of therapeutic approaches to activate innate reparative responses.


Asunto(s)
Riñón/citología , Riñón/fisiología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Separación Celular/métodos , Humanos , Células Madre Pluripotentes Inducidas , Riñón/crecimiento & desarrollo , Regeneración , Técnicas de Cultivo de Tejidos/métodos , Andamios del Tejido
14.
Proc Natl Acad Sci U S A ; 111(17): 6335-40, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24733901

RESUMEN

MicroRNAs (miRNAs) are major posttranscriptional regulators of a wide variety of biological processes. However, redundancy among most miRNAs has made it difficult to identify their in vivo functions. We previously demonstrated that global inhibition of miRNA biogenesis in Xenopus resulted in a dramatically smaller pronephric kidney. This suggested that microRNAs play a pivotal role in organ size control. Here we now provide a detailed mechanistic explanation for this phenotype. We identified that the activation of the mechanistic target of rapamycin complex 1 (mTORC1) by Insulin and insulin-like growth factor (Igf) 2 is an important regulator in kidney growth, which in turn is modulated by microRNAs. Molecular analyses demonstrate that microRNAs set a threshold for mTORC1 signaling by down-regulating one of its core negative regulators, tuberous sclerosis 1 (Tsc1). Most importantly, this rheostat can be reprogrammed experimentally. Whereas knockdown of miRNAs causes growth arrest, concomitant knockdown of Tsc1 restores mTORC1 activity and proximal tubular size. Together, these data establish a previously unidentified in vivo paradigm for the importance of posttranscriptional regulation in organ size control.


Asunto(s)
Riñón/anatomía & histología , MicroARNs/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Xenopus/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Riñón/embriología , Riñón/metabolismo , Túbulos Renales Proximales/anatomía & histología , Túbulos Renales Proximales/embriología , Túbulos Renales Proximales/metabolismo , Células LLC-PK1 , Diana Mecanicista del Complejo 1 de la Rapamicina , MicroARNs/genética , Tamaño de los Órganos/genética , Transducción de Señal/genética , Porcinos , Proteína 1 del Complejo de la Esclerosis Tuberosa , Xenopus/embriología
15.
J Am Soc Nephrol ; 27(4): 1159-73, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26311459

RESUMEN

Mutations in polycystin-1 (PC1) give rise to autosomal dominant polycystic kidney disease, an important and common cause of kidney failure. Despite its medical importance, the function of PC1 remains poorly understood. Here, we investigated the role of the intracellular polycystin-1, lipoxygenase, and α-toxin (PLAT) signature domain of PC1 using nuclear magnetic resonance, biochemical, cellular, and in vivo functional approaches. We found that the PLAT domain targets PC1 to the plasma membrane in polarized epithelial cells by a mechanism involving the selective binding of the PLAT domain to phosphatidylserine and L-α-phosphatidylinositol-4-phosphate (PI4P) enriched in the plasma membrane. This process is regulated by protein kinase A phosphorylation of the PLAT domain, which reduces PI4P binding and recruits ß-arrestins and the clathrin adaptor AP2 to trigger PC1 internalization. Our results reveal a physiological role for the PC1-PLAT domain in renal epithelial cells and suggest that phosphorylation-dependent internalization of PC1 is closely linked to its function in renal development and homeostasis.


Asunto(s)
Lipooxigenasa/fisiología , Canales Catiónicos TRPP/fisiología , Humanos , Lipooxigenasa/genética , Mutación , Estructura Terciaria de Proteína , Canales Catiónicos TRPP/genética
16.
Dev Dyn ; 245(8): 854-73, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27144987

RESUMEN

BACKGROUND: CUG-BP, Elav-like family member 1 (CELF1) is a multifunctional RNA binding protein found in a variety of adult and embryonic tissues. In the heart, CELF1 is found exclusively in the myocardium. However, the roles of CELF1 during cardiac development have not been completely elucidated. RESULTS: Myofibrillar organization is disrupted and proliferation is reduced following knockdown of CELF1 in cultured chicken primary embryonic cardiomyocytes. In vivo knockdown of Celf1 in developing Xenopus laevis embryos resulted in myofibrillar disorganization and a trend toward reduced proliferation in heart muscle, indicating conserved roles for CELF1 orthologs in embryonic cardiomyocytes. Loss of Celf1 also resulted in morphogenetic abnormalities in the developing heart and gut. Using optical coherence tomography, we showed that cardiac contraction was impaired following depletion of Celf1, while heart rhythm remained unperturbed. In contrast to cardiac muscle, loss of Celf1 did not disrupt myofibril organization in skeletal muscle cells, although it did lead to fragmentation of skeletal muscle bundles. CONCLUSIONS: CELF1 is required for normal myofibril organization, proliferation, morphogenesis, and contractile performance in the developing myocardium. Developmental Dynamics 245:854-873, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas CELF1/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Empalme Alternativo/genética , Animales , Western Blotting , Proteínas CELF1/genética , Células Cultivadas , Embrión de Pollo , Corazón/embriología , Inmunohistoquímica , Morfogénesis/genética , Morfogénesis/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Xenopus laevis
17.
Development ; 140(14): 3008-17, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23760952

RESUMEN

Syndecan 4 (Sdc4) is a cell-surface heparan sulfate proteoglycan (HSPG) that regulates gastrulation, neural tube closure and directed neural crest migration in Xenopus development. To determine whether Sdc4 participates in Wnt/PCP signaling during mouse development, we evaluated a possible interaction between a null mutation of Sdc4 and the loop-tail allele of Vangl2. Sdc4 is expressed in multiple tissues, but particularly in the non-neural ectoderm, hindgut and otic vesicles. Sdc4;Vangl2(Lp) compound mutant mice have defective spinal neural tube closure, disrupted orientation of the stereocilia bundles in the cochlea and delayed wound healing, demonstrating a strong genetic interaction. In Xenopus, co-injection of suboptimal amounts of Sdc4 and Vangl2 morpholinos resulted in a significantly greater proportion of embryos with defective neural tube closure than each individual morpholino alone. To probe the mechanism of this interaction, we overexpressed or knocked down Vangl2 function in HEK293 cells. The Sdc4 and Vangl2 proteins colocalize, and Vangl2, particularly the Vangl2(Lp) mutant form, diminishes Sdc4 protein levels. Conversely, Vangl2 knockdown enhances Sdc4 protein levels. Overall HSPG steady-state levels were regulated by Vangl2, suggesting a molecular mechanism for the genetic interaction in which Vangl2(Lp/+) enhances the Sdc4-null phenotype. This could be mediated via heparan sulfate residues, as Vangl2(Lp/+) embryos fail to initiate neural tube closure and develop craniorachischisis (usually seen only in Vangl2(Lp/Lp)) when cultured in the presence of chlorate, a sulfation inhibitor. These results demonstrate that Sdc4 can participate in the Wnt/PCP pathway, unveiling its importance during neural tube closure in mammalian embryos.


Asunto(s)
Polaridad Celular , Embrión de Mamíferos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Tubo Neural/citología , Sindecano-4/metabolismo , Vía de Señalización Wnt , Animales , Embrión de Mamíferos/citología , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Células Ciliadas Auditivas/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Ratones , Proteínas del Tejido Nervioso/genética , Tubo Neural/metabolismo , Defectos del Tubo Neural/metabolismo , Sindecano-4/genética , Cicatrización de Heridas , Xenopus
18.
Cell Mol Life Sci ; 72(12): 2415-29, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25650235

RESUMEN

Dysfunction of many ciliary proteins has been linked to a list of diseases, from cystic kidney to obesity and from hypertension to mental retardation. We previously proposed that primary cilia are unique communication organelles that function as microsensory compartments that house mechanosensory molecules. Here we report that primary cilia exhibit membrane swellings or ciliary bulbs, which based on their unique ultrastructure and motility, could be mechanically regulated by fluid-shear stress. Together with the ultrastructure analysis of the swelling, which contains monosialodihexosylganglioside (GM3), our results show that ciliary bulb has a distinctive set of functional proteins, including GM3 synthase (GM3S), bicaudal-c1 (Bicc1), and polycystin-2 (PC2). In fact, results from our cilia isolation demonstrated for the first time that GM3S and Bicc1 are members of the primary cilia proteins. Although these proteins are not required for ciliary membrane swelling formation under static condition, fluid-shear stress induced swelling formation is partially modulated by GM3S. We therefore propose that the ciliary bulb exhibits a sensory function within the mechano-ciliary structure. Overall, our studies provided an important step towards understanding the ciliary bulb function and structure.


Asunto(s)
Membrana Celular/fisiología , Cilios/fisiología , Células Epiteliales/metabolismo , Riñón/metabolismo , Mecanotransducción Celular/fisiología , Proteínas de Unión al ARN/metabolismo , Sialiltransferasas/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Células Epiteliales/citología , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Riñón/citología , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Sialiltransferasas/antagonistas & inhibidores , Sialiltransferasas/genética , Porcinos , Canales Catiónicos TRPP/antagonistas & inhibidores , Canales Catiónicos TRPP/genética
19.
Dev Biol ; 394(1): 54-64, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25127994

RESUMEN

The kidney is a homeostatic organ required for waste excretion and reabsorption of water, salts and other macromolecules. To this end, a complex series of developmental steps ensures the formation of a correctly patterned and properly proportioned organ. While previous studies have mainly focused on the individual signaling pathways, the formation of higher order receptor complexes in lipid rafts is an equally important aspect. These membrane platforms are characterized by differences in local lipid and protein compositions. Indeed, the cells in the Xenopus pronephric kidney were positive for the lipid raft markers ganglioside GM1 and Caveolin-1. To specifically interfere with lipid raft function in vivo, we focused on the Sterol Carrier Protein 2 (scp2), a multifunctional protein that is an important player in remodeling lipid raft composition. In Xenopus, scp2 mRNA was strongly expressed in differentiated epithelial structures of the pronephric kidney. Knockdown of scp2 did not interfere with the patterning of the kidney along its proximo-distal axis, but dramatically decreased the size of the kidney, in particular the proximal tubules. This phenotype was accompanied by a reduction of lipid rafts, but was independent of the peroxisomal or transcriptional activities of scp2. Finally, disrupting lipid micro-domains by inhibiting cholesterol synthesis using Mevinolin phenocopied the defects seen in scp2 morphants. Together these data underscore the importance for localized signaling platforms in the proper formation of the Xenopus kidney.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Túbulos Renales Proximales/embriología , Microdominios de Membrana/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Anticolesterolemiantes/farmacología , Tipificación del Cuerpo/genética , Línea Celular , Colesterol/biosíntesis , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Túbulos Renales Proximales/fisiología , Lovastatina/farmacología , Microdominios de Membrana/fisiología , Morfolinos , ARN Mensajero/biosíntesis , Transcripción Genética
20.
Hum Mol Genet ; 22(10): 1924-39, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23390129

RESUMEN

Mutations in PKD1 (85%) or PKD2 (15%) account for almost all cases of autosomal dominant polycystic kidney disease (ADPKD). The ADPKD proteins, termed as polycystin-1 (PC1) and polycystin-2 (PC2), interact via their C-termini to form a receptor-ion channel complex whose function and regulation are not fully understood. Here, we report the first phosphorylated residue (Ser(829)) in PC2, whose dephosphorylation is mediated by PC1 binding through the recruitment of protein phosphatase-1 alpha (PP1α). Using a new phosphospecific antibody (pPC2) to this site, we demonstrate that Ser(829) is phosphorylated by Protein kinase A (PKA) but remains constitutively phosphorylated in cells and tissues lacking PC1. cAMP increased pSer(829) basolateral localization in MDCK cells in a time dependent manner and was essential for pronephric development in Xenopus embryos. When constitutively expressed, a complex phenotype associated with enhanced ATP-dependent ER Ca(2+) release and loss of growth suppression was observed in cycling cells. These results reveal a reciprocal functional link between PC1 and PC2 which is critically dependent on their interaction. Unopposed cAMP stimulated hyperphosphorylation of PC2 in the absence of functional PC1 could contribute to cyst initiation in PKD1 patients and represents a new molecular paradigm in understanding ADPKD pathogenesis.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteína Fosfatasa 1/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Perros , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Mutación , Fosforilación/fisiología , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Proteína Fosfatasa 1/genética , Estructura Terciaria de Proteína , Canales Catiónicos TRPP/genética , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA